Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
2.
Stem Cell Reports ; 10(1): 212-227, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29249663

ABSTRACT

Here, we show that HEMATOLOGICAL AND NEUROLOGICAL EXPRESSED 1-LIKE (HN1L) is a targetable breast cancer stem cell (BCSC) gene that is altered in 25% of whole breast cancer and significantly correlated with shorter overall or relapse-free survival in triple-negative breast cancer (TNBC) patients. HN1L silencing reduced the population of BCSCs, inhibited tumor initiation, resensitized chemoresistant tumors to docetaxel, and hindered cancer progression in multiple TNBC cell line-derived xenografts. Additionally, gene signatures associated with HN1L correlated with shorter disease-free survival of TNBC patients. We defined HN1L as a BCSC transcription regulator for genes involved in the LEPR-STAT3 signaling axis as HN1L binds to a putative consensus upstream sequence of STAT3, LEPTIN RECEPTOR, and MIR-150. Our data reveal that BCSCs in TNBC depend on the transcription regulator HN1L for the sustained activation of the LEPR-STAT3 pathway, which makes it a potentially important target for both prognosis and BCSC therapy.


Subject(s)
Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Receptors, Leptin/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Triple Negative Breast Neoplasms/metabolism , Animals , Cell Line, Tumor , Female , Humans , Mice , Mice, SCID , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Proteins/genetics , Neoplastic Stem Cells/pathology , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Receptors, Leptin/genetics , Response Elements , STAT3 Transcription Factor/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
3.
J Biol Chem ; 291(45): 23756-23768, 2016 Nov 04.
Article in English | MEDLINE | ID: mdl-27650498

ABSTRACT

Bromodomain and extraterminal (BET) proteins are epigenetic "readers" that recognize acetylated histones and mark areas of the genome for transcription. BRD4, a BET family member protein, has been implicated in a number of types of cancer, and BET protein inhibitors (BETi) are efficacious in many preclinical cancer models. However, the drivers of response to BETi vary depending on tumor type, and little is known regarding the target genes conveying BETi activity in triple-negative breast cancer (TNBC). Here, we show that BETi repress growth of multiple in vitro and in vivo models of TNBC by inducing two terminal responses: apoptosis and senescence. Unlike in other cancers, response to BETi in TNBC is not dependent upon suppression of MYC Instead, both end points are preceded by the appearance of polyploid cells caused by the suppression of Aurora kinases A and B (AURKA/B), which are critical mediators of mitosis. In addition, AURKA/B inhibitors phenocopy the effects of BETi. These results indicate that Aurora kinases play an important role in the growth suppressive activity of BETi in TNBC. Elucidating the mechanism of response to BETi in TNBC should 1) facilitate the prediction of how distinct TNBC tumors will respond to BETi and 2) inform the rational design of drug combination therapies.


Subject(s)
Antineoplastic Agents/therapeutic use , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase B/antagonists & inhibitors , Breast/drug effects , Nuclear Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Transcription Factors/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Aurora Kinase A/metabolism , Aurora Kinase B/metabolism , Breast/metabolism , Breast/pathology , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Nuclear Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
4.
Sci Transl Med ; 8(334): 334ra53, 2016 Apr 13.
Article in English | MEDLINE | ID: mdl-27075627

ABSTRACT

Amplifications at 9p24 have been identified in breast cancer and other malignancies, but the genes within this locus causally associated with oncogenicity or tumor progression remain unclear. Targeted next-generation sequencing of postchemotherapy triple-negative breast cancers (TNBCs) identified a group of 9p24-amplified tumors, which contained focal amplification of the Janus kinase 2 (JAK2) gene. These patients had markedly inferior recurrence-free and overall survival compared to patients with TNBC without JAK2 amplification. Detection of JAK2/9p24 amplifications was more common in chemotherapy-treated TNBCs than in untreated TNBCs or basal-like cancers, or in other breast cancer subtypes. Similar rates of JAK2 amplification were confirmed in patient-derived TNBC xenografts. In patients for whom longitudinal specimens were available, JAK2 amplification was selected for during neoadjuvant chemotherapy and eventual metastatic spread, suggesting a role in tumorigenicity and chemoresistance, phenotypes often attributed to a cancer stem cell-like cell population. In TNBC cell lines with JAK2 copy gains or amplification, specific inhibition of JAK2 signaling reduced mammosphere formation and cooperated with chemotherapy in reducing tumor growth in vivo. In these cells, inhibition of JAK1-signal transducer and activator of transcription 3 (STAT3) signaling had little effect or, in some cases, counteracted JAK2-specific inhibition. Collectively, these results suggest that JAK2-specific inhibitors are more efficacious than dual JAK1/2 inhibitors against JAK2-amplified TNBCs. Furthermore, JAK2 amplification is a potential biomarker for JAK2 dependence, which, in turn, can be used to select patients for clinical trials with JAK2 inhibitors.


Subject(s)
Chromosomes, Human, Pair 9/genetics , Gene Amplification , Genetic Loci , Janus Kinase 2/genetics , Triple Negative Breast Neoplasms/enzymology , Triple Negative Breast Neoplasms/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Cohort Studies , Female , Gene Knockdown Techniques , Humans , Middle Aged , STAT3 Transcription Factor/metabolism , STAT6 Transcription Factor/metabolism , Signal Transduction/drug effects , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology
5.
Breast Cancer Res ; 18(1): 6, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26757880

ABSTRACT

BACKGROUND: Developing novel strategies against treatment-resistant triple negative breast cancer (TNBC) cells remains a significant challenge. The ErbB family, including epidermal growth factor receptor (EGFR), plays key roles in metastasis, tumorigenesis, cell proliferation, and drug resistance. Recently, these characteristics have been linked to a small subpopulation of cells classified as cancer stem cells (CSC) which are believed to be responsible for tumor initiation and maintenance. Ixabepilone is a new generation microtubule-stabilizing agent, which has been expected to be more efficacious than conventional taxanes. Here we aim to investigate whether the EGFR monoclonal antibody Cetuximab, in combination with Ixabepilone, is more effective in eliminating CSC populations compared to chemotherapy alone in TNBC. METHODS: Representative TNBC cell lines (MDA-MB-231 and SUM159) were used to evaluate breast CSC populations. We used fluorescence-activated cell sorter analysis (CD44(+) and CD24(-/low), or Aldefluor(+)) and a self-renewal assay called mammosphere formation efficiency (MSFE) to measure CSC population size after treatment with Cetuximab, or Cetuximab plus Ixabepilone in vitro. RESULTS: Although there was no significant decrease in cell viability, Cetuximab reduced MSFE and the CSC population in breast cancer cells in vitro and in vivo through inhibition of autophagy. Also, SUM159 and MDA-MB-231 orthotopic tumors demonstrated partial response to Centuximab or Ixabepilone monotherapy; however, the effect of the combination treatment was significant only in SUM159 tumors (p <0.0001), when compared to Ixabepilone alone. CONCLUSIONS: Overall, our findings demonstrate that EGFR-targeted therapy by Cetuximab effectively reduces the CSC population in TNBC tumors. However, combination therapy with Ixabepilone may be effective only in a small subset of TNBCs, warranting further investigation of alternative approaches to target multiple pathways for TNBC treatment.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cetuximab/administration & dosage , Epothilones/administration & dosage , Triple Negative Breast Neoplasms/drug therapy , Antibodies, Monoclonal, Humanized/administration & dosage , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Female , Humans , Neoplastic Stem Cells/drug effects , Triple Negative Breast Neoplasms/pathology
6.
Breast Cancer Res ; 17: 25, 2015 Feb 22.
Article in English | MEDLINE | ID: mdl-25849745

ABSTRACT

INTRODUCTION: Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer with no effective targeted therapy. Inducible nitric oxide synthase (iNOS) is associated with poor survival in patients with breast cancer by increasing tumor aggressiveness. This work aimed to investigate the potential of iNOS inhibitors as a targeted therapy for TNBC. We hypothesized that inhibition of endogenous iNOS would decrease TNBC aggressiveness by reducing tumor initiation and metastasis through modulation of epithelial-mesenchymal transition (EMT)-inducing factors. METHODS: iNOS protein levels were determined in 83 human TNBC tissues and correlated with clinical outcome. Proliferation, mammosphere-forming efficiency, migration, and EMT transcription factors were assessed in vitro after iNOS inhibition. Endogenous iNOS targeting was evaluated as a potential therapy in TNBC mouse models. RESULTS: High endogenous iNOS expression was associated with worse prognosis in patients with TNBC by gene expression as well as immunohistochemical analysis. Selective iNOS (1400 W) and pan-NOS (L-NMMA and L-NAME) inhibitors diminished cell proliferation, cancer stem cell self-renewal, and cell migration in vitro, together with inhibition of EMT transcription factors (Snail, Slug, Twist1, and Zeb1). Impairment of hypoxia-inducible factor 1α, endoplasmic reticulum stress (IRE1α/XBP1), and the crosstalk between activating transcription factor 3/activating transcription factor 4 and transforming growth factor ß was observed. iNOS inhibition significantly reduced tumor growth, the number of lung metastases, tumor initiation, and self-renewal. CONCLUSIONS: Considering the effectiveness of L-NMMA in decreasing tumor growth and enhancing survival rate in TNBC, we propose a targeted therapeutic clinical trial by re-purposing the pan-NOS inhibitor L-NMMA, which has been extensively investigated for cardiogenic shock as an anti-cancer therapeutic.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Triple Negative Breast Neoplasms/metabolism , Activating Transcription Factor 3/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Endoplasmic Reticulum Stress , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung Neoplasms/secondary , Mice , Molecular Targeted Therapy , Neoplasm Invasiveness , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Prognosis , Transforming Growth Factor beta/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
7.
Stem Cells ; 32(9): 2309-23, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24809620

ABSTRACT

Triple negative breast cancer (TNBC) is known to contain a high percentage of CD44(+) /CD24(-/low) cancer stem cells (CSCs), corresponding with a poor prognosis despite systemic chemotherapy. Chloroquine (CQ), an antimalarial drug, is a lysotropic reagent which inhibits autophagy. CQ was identified as a potential CSC inhibitor through in silico gene expression signature analysis of the CD44(+) /CD24(-/low) CSC population. Autophagy plays a critical role in adaptation to stress conditions in cancer cells, and is related with drug resistance and CSC maintenance. Thus, the objectives of this study were to examine the potential enhanced efficacy arising from addition of CQ to standard chemotherapy (paclitaxel) in TNBC and to identify the mechanism by which CQ eliminates CSCs in TNBCs. Herein, we report that CQ sensitizes TNBC cells to paclitaxel through inhibition of autophagy and reduces the CD44(+) /CD24(-/low) CSC population in both preclinical and clinical settings. Also, we are the first to report a mechanism by which CQ regulates the CSCs in TNBC through inhibition of the Janus-activated kinase 2 (Jak2)-signal transducer and activator of transcription 3 signaling pathway by reducing the expression of Jak2 and DNA methyltransferase 1.


Subject(s)
Chloroquine/pharmacology , DNA (Cytosine-5-)-Methyltransferases/metabolism , Janus Kinase 2/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Animals , Autophagy/drug effects , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferase 1 , Female , Humans , Mice , Mice, Nude , Neoplastic Stem Cells/pathology , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/metabolism
8.
Small ; 10(13): 2688-96, 2014 Jul 09.
Article in English | MEDLINE | ID: mdl-24639392

ABSTRACT

Hybrid PET/MRI scanners have the potential to provide fundamental molecular, cellular, and anatomic information essential for optimizing therapeutic and surgical interventions. However, their full utilization is currently limited by the lack of truly multi-modal contrast agents capable of exploiting the strengths of each modality. Here, we report on the development of long-circulating positron-emitting magnetic nanoconstructs (PEM) designed to image solid tumors for combined PET/MRI. PEMs are synthesized by a modified nano-precipitation method mixing poly(lactic-co-glycolic acid) (PLGA), lipids, and polyethylene glycol (PEG) chains with 5 nm iron oxide nanoparticles (USPIOs). PEM lipids are coupled with 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) and subsequently chelated to (64)Cu. PEMs show a diameter of 140 ± 7 nm and a transversal relaxivity r2 of 265.0 ± 10.0 (mM × s)(-1), with a r2/r1 ratio of 123. Using a murine xenograft model bearing human breast cancer cell line (MDA-MB-231), intravenously administered PEMs progressively accumulate in tumors reaching a maximum of 3.5 ± 0.25% ID/g tumor at 20 h post-injection. Correlation of PET and MRI signals revealed non-uniform intratumoral distribution of PEMs with focal areas of accumulation at the tumor periphery. These long-circulating PEMs with high transversal relaxivity and tumor accumulation may allow for detailed interrogation over multiple scales in a clinically relevant setting.


Subject(s)
Electrons , Magnetic Resonance Imaging , Magnetics , Positron-Emission Tomography , Animals , Mice , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/pathology
9.
Cell Res ; 24(5): 542-59, 2014 May.
Article in English | MEDLINE | ID: mdl-24675532

ABSTRACT

Combinatorial targeted therapies are more effective in treating cancer by blocking by-pass mechanisms or inducing synthetic lethality. However, their clinical application is hampered by resistance and toxicity. To meet this important challenge, we developed and tested a novel concept of biomarker-guided sequential applications of various targeted therapies using ErbB2-overexpressing/PTEN-low, highly aggressive breast cancer as our model. Strikingly, sustained activation of ErbB2 and downstream pathways drives trastuzumab resistance in both PTEN-low/trastuzumab-resistant breast cancers from patients and mammary tumors with intratumoral heterogeneity from genetically-engineered mice. Although lapatinib initially inhibited trastuzumab-resistant mouse tumors, tumors by-passed the inhibition by activating the PI3K/mTOR signaling network as shown by the quantitative protein arrays. Interestingly, activation of the mTOR pathway was also observed in neoadjuvant lapatinib-treated patients manifesting lapatinib resistance. Trastuzumab + lapatinib resistance was effectively overcome by sequential application of a PI3K/mTOR dual kinase inhibitor (BEZ235) with no significant toxicity. However, our p-RTK array analysis demonstrated that BEZ235 treatment led to increased ErbB2 expression and phosphorylation in genetically-engineered mouse tumors and in 3-D, but not 2-D, culture, leading to BEZ235 resistance. Mechanistically, we identified ErbB2 protein stabilization and activation as a novel mechanism of BEZ235 resistance, which was reversed by subsequent treatment with lapatinib + BEZ235 combination. Remarkably, this sequential application of targeted therapies guided by biomarker changes in the tumors rapidly evolving resistance doubled the life-span of mice bearing exceedingly aggressive tumors. This fundamentally novel approach of using targeted therapies in a sequential order can effectively target and reprogram the signaling networks in cancers evolving resistance during treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast/drug effects , Drug Resistance, Neoplasm/drug effects , Molecular Targeted Therapy/methods , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Humans , Imidazoles/pharmacology , Lapatinib , Mice , Mice, Transgenic , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Quinolines/pharmacology , Receptor, ErbB-2/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Trastuzumab , Tumor Cells, Cultured
10.
Nature ; 508(7494): 103-107, 2014 Apr 03.
Article in English | MEDLINE | ID: mdl-24670641

ABSTRACT

Cancer cells induce a set of adaptive response pathways to survive in the face of stressors due to inadequate vascularization. One such adaptive pathway is the unfolded protein (UPR) or endoplasmic reticulum (ER) stress response mediated in part by the ER-localized transmembrane sensor IRE1 (ref. 2) and its substrate XBP1 (ref. 3). Previous studies report UPR activation in various human tumours, but the role of XBP1 in cancer progression in mammary epithelial cells is largely unknown. Triple-negative breast cancer (TNBC)--a form of breast cancer in which tumour cells do not express the genes for oestrogen receptor, progesterone receptor and HER2 (also called ERBB2 or NEU)--is a highly aggressive malignancy with limited treatment options. Here we report that XBP1 is activated in TNBC and has a pivotal role in the tumorigenicity and progression of this human breast cancer subtype. In breast cancer cell line models, depletion of XBP1 inhibited tumour growth and tumour relapse and reduced the CD44(high)CD24(low) population. Hypoxia-inducing factor 1α (HIF1α) is known to be hyperactivated in TNBCs. Genome-wide mapping of the XBP1 transcriptional regulatory network revealed that XBP1 drives TNBC tumorigenicity by assembling a transcriptional complex with HIF1α that regulates the expression of HIF1α targets via the recruitment of RNA polymerase II. Analysis of independent cohorts of patients with TNBC revealed a specific XBP1 gene expression signature that was highly correlated with HIF1α and hypoxia-driven signatures and that strongly associated with poor prognosis. Our findings reveal a key function for the XBP1 branch of the UPR in TNBC and indicate that targeting this pathway may offer alternative treatment strategies for this aggressive subtype of breast cancer.


Subject(s)
DNA-Binding Proteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Animals , CD24 Antigen/metabolism , Cell Hypoxia/genetics , Cell Line, Tumor , Cell Proliferation , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Gene Silencing , Humans , Hyaluronan Receptors/metabolism , Mice , Neoplasm Invasiveness , Neoplasm Recurrence, Local , Prognosis , RNA Polymerase II/metabolism , Regulatory Factor X Transcription Factors , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription, Genetic , Triple Negative Breast Neoplasms/blood supply , Triple Negative Breast Neoplasms/genetics , Unfolded Protein Response , X-Box Binding Protein 1
11.
Stem Cell Reports ; 2(1): 78-91, 2014 Jan 14.
Article in English | MEDLINE | ID: mdl-24511467

ABSTRACT

Previous studies have suggested that breast cancer stem cells (BCSCs) mediate metastasis, are resistant to radiation and chemotherapy, and contribute to relapse. Although several BCSC markers have been described, it is unclear whether these markers identify the same or independent BCSCs. Here, we show that BCSCs exist in distinct mesenchymal-like (epithelial-mesenchymal transition [EMT]) and epithelial-like (mesenchymal-epithelial transition [MET]) states. Mesenchymal-like BCSCs characterized as CD24(-)CD44(+) are primarily quiescent and localized at the tumor invasive front, whereas epithelial-like BCSCs express aldehyde dehydrogenase (ALDH), are proliferative, and are located more centrally. The gene-expression profiles of mesenchymal-like and epithelial-like BCSCs are remarkably similar across different molecular subtypes of breast cancer, and resemble those of distinct basal and luminal stem cells found in the normal breast. We propose that the plasticity of BCSCs that allows them to transition between EMT- and MET-like states endows these cells with the capacity for tissue invasion, dissemination, and growth at metastatic sites.


Subject(s)
Epithelial-Mesenchymal Transition , Neoplastic Stem Cells/cytology , Aldehyde Dehydrogenase/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CD24 Antigen/metabolism , Epithelial Cells/cytology , Female , Humans , Hyaluronan Receptors/metabolism , MCF-7 Cells , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Mesenchymal Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Transcriptome
12.
New J Phys ; 15: 55004, 2013 May 08.
Article in English | MEDLINE | ID: mdl-24039540

ABSTRACT

Heterogeneities in the perfusion of solid tumors prevent optimal delivery of nanotherapeutics. Clinical imaging protocols to obtain patient-specific data have proven difficult to implement. It is challenging to determine which perfusion features hold greater prognostic value and to relate measurements to vessel structure and function. With the advent of systemically administered nanotherapeutics, whose delivery is dependent on overcoming diffusive and convective barriers to transport, such knowledge is increasingly important. We describe a framework for the automated evaluation of vascular perfusion curves measured at the single vessel level. Primary tumor fragments, collected from triple-negative breast cancer patients and grown as xenografts in mice, were injected with fluorescence contrast and monitored using intravital microscopy. The time to arterial peak and venous delay, two features whose probability distributions were measured directly from time-series curves, were analyzed using a Fuzzy C-mean (FCM) supervised classifier in order to rank individual tumors according to their perfusion characteristics. The resulting rankings correlated inversely with experimental nanoparticle accumulation measurements, enabling modeling of nanotherapeutics delivery without requiring any underlying assumptions about tissue structure or function, or heterogeneities contained within. With additional calibration, these methodologies may enable the study of nanotherapeutics delivery strategies in a variety of tumor models.

13.
Breast Cancer Res ; 15(5): R77, 2013.
Article in English | MEDLINE | ID: mdl-24008095

ABSTRACT

INTRODUCTION: We hypothesized that cells present in normal tissue that bear cancer stem cell markers may represent a cancer cell of origin or a microenvironment primed for tumor development, and that their presence may correlate with the clinically defined subtypes of breast cancer that show increased tumorigenicity and stem cell features. methods: Normal tissues sampled at least 5 cm from primary tumors (normal adjacent tissue) were obtained from 61 chemotherapy-naive patients with breast cancer treated with mastectomy. Samples were stained simultaneously with immunofluorescence for CD44/CD49f/CD133/2 stem cell markers. We assessed the association between CD44+CD49f+CD133/2+ staining in normal adjacent tissue and breast cancer receptor subtype (defined by the expression of the estrogen (ER), progesterone (PR), or human epidermal growth factor-2 (Her2) receptors). We also examined the correlation between CD44+CD49f+CD133/2+ immunofluorescence and each of two previously published gene signatures, one derived from stem-cell enriched tissue and one from BRCA mutated tissue expected to have defective DNA repair. RESULTS: Patients with triple negative breast cancer (ER­/PR­/HER2­) expressed CD44+CD49f+CD133/2+ in 9 of 9 normal adjacent tissue samples compared with 7 of 52 ER+ and/or Her2+ tumors (P < 0.001). Further, expression of CD44+CD49f+CD133/2+ by normal adjacent tissue correlated positively with a stem cell-derived tumorigenic signature (P <0.001) and inversely with a defective DNA-repair signature (P <0.001). CONCLUSION: Normal cells bearing cancer stem cell markers are associated with the triple negative receptor subtype of breast cancer. This study suggests stem cell staining and gene expression signatures from normal breast tissues represent novel tissue-based risk biomarkers for triple negative breast cancer. Validation of these results in additional studies of normal tissue from cancer-free women could lay the foundation for future targeted triple negative breast cancer prevention strategies.


Subject(s)
DNA Repair , Neoplastic Stem Cells/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , AC133 Antigen , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Surface , Biomarkers/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Epidemiologic Factors , Female , Gene Expression Profiling , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Integrin alpha6/genetics , Integrin alpha6/metabolism , Mammary Glands, Human/metabolism , Peptides/genetics , Peptides/metabolism , Triple Negative Breast Neoplasms/mortality
14.
Cancer Res ; 73(15): 4885-97, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23737486

ABSTRACT

Breast cancer research is hampered by difficulties in obtaining and studying primary human breast tissue, and by the lack of in vivo preclinical models that reflect patient tumor biology accurately. To overcome these limitations, we propagated a cohort of human breast tumors grown in the epithelium-free mammary fat pad of severe combined immunodeficient (SCID)/Beige and nonobese diabetic (NOD)/SCID/IL-2γ-receptor null (NSG) mice under a series of transplant conditions. Both models yielded stably transplantable xenografts at comparably high rates (∼21% and ∼19%, respectively). Of the conditions tested, xenograft take rate was highest in the presence of a low-dose estradiol pellet. Overall, 32 stably transplantable xenograft lines were established, representing 25 unique patients. Most tumors yielding xenografts were "triple-negative" [estrogen receptor (ER)-progesterone receptor (PR)-HER2+; n = 19]. However, we established lines from 3 ER-PR-HER2+ tumors, one ER+PR-HER2-, one ER+PR+HER2-, and one "triple-positive" (ER+PR+HER2+) tumor. Serially passaged xenografts show biologic consistency with the tumor of origin, are phenotypically stable across multiple transplant generations at the histologic, transcriptomic, proteomic, and genomic levels, and show comparable treatment responses as those observed clinically. Xenografts representing 12 patients, including 2 ER+ lines, showed metastasis to the mouse lung. These models thus serve as a renewable, quality-controlled tissue resource for preclinical studies investigating treatment response and metastasis.


Subject(s)
Breast Neoplasms/pathology , Cell Line, Tumor , Xenograft Model Antitumor Assays/methods , Animals , Female , Humans , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Phenotype
15.
Biomaterials ; 34(21): 5402-10, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23611451

ABSTRACT

The in vivo performance of nanoparticles is affected by their size, shape and surface properties. Fabrication methods based on emulsification and nano-precipitation cannot control these features precisely and independently over multiple scales. Herein, discoidal polymeric nanoconstructs (DPNs) with a diameter of 1000 nm and a height of 500 nm are demonstrated via a modified hydrogel-template strategy. The DPNs are obtained by mixing in one synthesis step the constituent polymers - poly(lactic acid-co-glycolic acid) (PLGA) and polyethylene glycol (PEG) dimethacrylate - and the payload with magneto-optical properties - 5 nm ultra-small super-paramagnetic iron oxide nanoparticles (SPIOs) and Rhodamine B dye (RhB). The DPN geometrical features are characterized by multiple microscopy techniques. The release of the Rhodamine B dye is pH dependent and increases under acidic conditions by the enhanced hydrolysis of the polymeric matrix. Each DPN is loaded with ~100 fg of iron and can be efficiently dragged by static and external magnetic fields. Moreover, the USPIO confinement within the DPN porous structure is responsible for a significant enhancement in MRI relaxivity (r2 ~ 500 (mMs)(-1)), up to ~5 times larger than commercially available systems. These nanoconstructs suggest a general strategy to engineer theranostic systems for anti-angiogenic treatment and vascular imaging.


Subject(s)
Diagnostic Imaging/methods , Lactic Acid/chemistry , Magnetic Phenomena , Methacrylates/chemistry , Nanotechnology/methods , Neoplasms/diagnosis , Optical Phenomena , Polyethylene Glycols/chemistry , Polyglycolic Acid/chemistry , Animals , Cell Death/drug effects , HeLa Cells , Humans , Mice , Mice, SCID , Microscopy, Fluorescence , Nanoparticles/toxicity , Nanoparticles/ultrastructure , Polylactic Acid-Polyglycolic Acid Copolymer , Rhodamines/metabolism
16.
Clin Cancer Res ; 19(6): 1512-24, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23340294

ABSTRACT

PURPOSE: Accumulating evidence supports the existence of breast cancer stem cells (BCSC), which are characterized by their capacity to self-renew and divide indefinitely and resistance to conventional therapies. The Notch pathway is important for stem cell renewal and is a potential target for BCSC-directed therapy. EXPERIMENTAL DESIGN: Using human breast tumorgraft studies, we evaluated the impact of gamma secretase inhibitors (GSI) on the BCSC population and the efficacy of combining GSI with docetaxel treatment. The mouse experimental therapy paralleled a concurrent clinical trial in patients with advanced breast cancer, designed to determine the maximum-tolerated dose of the GSI, MK-0752, administered sequentially with docetaxel, and to evaluate BCSC markers in serial tumor biopsies. RESULTS: Treatment with GSI reduced BCSCs in MC1 and BCM-2147 tumorgrafts by inhibition of the Notch pathway. GSI enhanced the efficacy of docetaxel in preclinical studies. In the clinical trial, 30 patients with advanced breast cancer were treated with escalating doses of MK-0752 plus docetaxel. Clinically, meaningful doses of both drugs were possible with manageable toxicity and preliminary evidence of efficacy. A decrease in CD44(+)/CD24(-), ALDH(+), and mammosphere-forming efficiency were observed in tumors of patients undergoing serial biopsies. CONCLUSIONS: These preclinical data show that pharmacologic inhibition of the Notch pathway can reduce BCSCs in breast tumorgraft models. The clinical trial shows feasibility of combination GSI and chemotherapy, and together these results encourage further study of Notch pathway inhibitors in combination with chemotherapy in breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Benzene Derivatives/administration & dosage , Breast Neoplasms/drug therapy , Propionates/administration & dosage , Sulfones/administration & dosage , Taxoids/administration & dosage , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Benzene Derivatives/adverse effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Docetaxel , Female , Humans , Maximum Tolerated Dose , Mice , Neoplasm Staging , Propionates/adverse effects , Receptors, Notch/metabolism , Signal Transduction , Sulfones/adverse effects , Taxoids/adverse effects , Xenograft Model Antitumor Assays
17.
Breast Cancer Res ; 15(1): 201, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23339383

ABSTRACT

Despite improved detection and reduction of breast cancer-related deaths over the recent decade, breast cancer remains the second leading cause of cancer death for women in the US, with 39,510 women expected to succumb to metastatic disease in 2012 alone (American Cancer Society, Cancer Facts &Figures 2012. Atlanta: American Cancer Society; 2012). Continued efforts in classification of breast cancers based on gene expression profiling and genomic sequencing have revealed an underlying complexity and molecular heterogeneity within the disease that continues to challenge therapeutic interventions. To successfully identify and translate new treatment regimens to the clinic, it is imperative that our preclinical models recapitulate this complexity and heterogeneity. In this review article, we discuss the recent advances in development and classification of patient-derived human breast tumor xenograft models that have the potential to facilitate the next phase of drug discovery for personalized cancer therapy based on the unique driver signaling pathways in breast tumor subtypes.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Precision Medicine , Xenograft Model Antitumor Assays , Breast Neoplasms/pathology , Female , Humans , Pathology, Molecular , Patients , United States
18.
Proc Natl Acad Sci U S A ; 109(46): 18707-12, 2012 Nov 13.
Article in English | MEDLINE | ID: mdl-23112172

ABSTRACT

Here we report a microfluidics method to enrich physically deformable cells by mechanical manipulation through artificial microbarriers. Driven by hydrodynamic forces, flexible cells or cells with high metastatic propensity change shape to pass through the microbarriers and exit the separation device, whereas stiff cells remain trapped. We demonstrate the separation of (i) a mixture of two breast cancer cell types (MDA-MB-436 and MCF-7) with distinct deformabilities and metastatic potentials, and (ii) a heterogeneous breast cancer cell line (SUM149), into enriched flexible and stiff subpopulations. We show that the flexible phenotype is associated with overexpression of multiple genes involved in cancer cell motility and metastasis, and greater mammosphere formation efficiency. Our observations support the relationship between tumor-initiating capacity and cell deformability, and demonstrate that tumor-initiating cells are less differentiated in terms of cell biomechanics.


Subject(s)
Cell Separation/instrumentation , Cell Separation/methods , Microfluidic Analytical Techniques , Neoplastic Stem Cells/cytology , Spheroids, Cellular/cytology , Cell Line, Tumor , Humans
19.
PLoS One ; 7(8): e30207, 2012.
Article in English | MEDLINE | ID: mdl-22879872

ABSTRACT

Metastasis and disease relapse are hypothesized to result from tumor initiating cells (TICs). Previously, we have defined a CD44+/CD24-/low mammosphere-forming tumorigenic 493-gene signature in breast cancer. Stat3 was identified as a critical node in self-renewal based on an ongoing lentiviral shRNA screen being conducted in two breast cancer cell lines SUM159 and BT549. In corroborating work, targeting the SH2 domain of Stat3 with a novel small molecule decreased the percentage of cells expressing TIC markers (CD44+/CD24-/low and ALDH+) and mammosphere formation in p-Stat3 overexpressing human breast cancer xenografts in SCID-beige mice. Importantly, we observed a four-fold improvement in the 30-day recurrence-free survival relative to docetaxel alone with the addition of the Stat3 inhibitor in the chemoresistant tumor model. Thus, these findings provide a strong impetus for the development of selective Stat3 inhibitors in order to improve survival in patients with p-Stat3 overexpressing tumors.


Subject(s)
Breast Neoplasms/pathology , Neoplastic Stem Cells/pathology , STAT3 Transcription Factor/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Xenograft Model Antitumor Assays , Adipose Tissue/drug effects , Animals , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Disease-Free Survival , Docetaxel , Drug Resistance, Neoplasm/drug effects , Female , Humans , Kaplan-Meier Estimate , Mice , Mice, SCID , Neoplasm Recurrence, Local , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , STAT3 Transcription Factor/metabolism , Small Molecule Libraries/therapeutic use , Taxoids/pharmacology , Taxoids/therapeutic use , Time Factors , Tumor Burden/drug effects
20.
Breast Cancer Res Treat ; 132(3): 993-1000, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21750966

ABSTRACT

Obesity is thought to contribute to worse disease outcome in breast cancer as a result of increased levels of adipocyte-secreted endocrine factors, insulin, and insulin-like growth factors (IGFs) that accelerate tumor cell proliferation and impair treatment response. We examined the effects of patient obesity on primary breast tumor gene expression, by profiling transcription of a set of 103 tumors for which the patients' body mass index (BMI) was ascertained. Sample profiles were stratified according to patients' obesity phenotype defined as normal (BMI < 25), overweight (BMI 25-29.9), or obese (BMI ≥ 30). Widespread gene expression alterations were evident in breast tumors from obese patients as compared to other tumors, allowing us to define an obesity-associated cancer transcriptional signature of 662 genes. In multiple public expression data sets of breast cancers (representing > 1,500 patients), manifestation of the obesity signature patterns correlated with manifestation of a gene signature for IGF signaling and (to a lesser extent) with lower levels of estrogen receptor. In one patient cohort, manifestation of the obesity signature correlated with shorter time to metastases. A number of small molecules either induced or suppressed the obesity-associated transcriptional program in vitro; estrogens alpha-estradiol, levonorgestrel, and hexestrol induced the program, while several anti-parkinsonian agents targeting neurotransmitter receptor pathways repressed the program. Obesity in breast cancer patients appears to impact the gene expression patterns of the tumor (perhaps as a result of altered body chemistry). These results warrant further investigation of obesity-associated modifiers of breast cancer risk and disease outcome.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/metabolism , Gene Expression Profiling , Obesity/metabolism , Biomarkers, Tumor/metabolism , Breast Neoplasms/complications , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Insulin-Like Growth Factor I/physiology , Kaplan-Meier Estimate , Obesity/complications , Obesity/genetics , Oligonucleotide Array Sequence Analysis , Proportional Hazards Models , Prospective Studies , Receptors, Estrogen/metabolism , Reference Values , Signal Transduction , Statistics, Nonparametric , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...